Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Kidney Int ; 72(5): 529-30, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17713560

ABSTRACT

The study by Rieg et al. is the first to examine potassium handling in BK(-/-) mice, thereby addressing many unanswered questions regarding the separate roles of BK and ROMK channels in renal potassium secretion. This Commentary is an interpretation and opinion of their results, by two researchers who have been studying BK and ROMK, respectively, as potassium secretory channels in the distal nephron.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Potassium Channels, Inwardly Rectifying/physiology , Animals , Mice , Mice, Knockout , Nephrons/chemistry , Potassium/urine
2.
Article in English | MEDLINE | ID: mdl-15674648

ABSTRACT

Exciting discoveries in the last decade have cast light onto the fundamental mechanisms that underlie polarized trafficking in epithelial cells. It is now clear that epithelial cell membrane asymmetry is achieved by a combination of intracellular sorting operations, vectorial delivery mechanisms and plasmalemma-specific fusion and retention processes. Several well-defined signals that specify polarized segregation, sorting, or retention processes have, now, been described in a number of proteins. The intracellular machineries that decode and act on these signals are beginning to be described. In addition, the nature of the molecules that associate with intracellular trafficking vesicles to coordinate polarized delivery, tethering, docking, and fusion are also becoming understood. Combined with direct visualization of polarized sorting processes with new technologies in live-cell fluorescent microscopy, new and surprising insights into these once-elusive trafficking processes are emerging. Here we provide a review of these recent advances within an historically relevant context.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/physiology , Kidney/cytology , Membrane Proteins/biosynthesis , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Kidney/physiology , Signal Transduction/physiology
3.
Proc Natl Acad Sci U S A ; 98(18): 10475-80, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504929

ABSTRACT

The inwardly rectifying potassium channel Kir 2.3 is specifically targeted and expressed on the basolateral membrane of certain renal epithelial cells. In the present study, the structural basis for polarized targeting was elucidated. Deletion of a unique COOH-terminal domain produced channels that were mistargeted to the apical membrane, consistent with the removal of a basolateral membrane-sorting signal. By characterizing a series of progressively smaller truncation mutants, an essential targeting signal was defined (residues 431-442) within a domain that juxtaposes or overlaps with a type I PDZ binding motif (442). Fusion of the COOH-terminal structure onto CD4 was sufficient to change a random membrane-trafficking and expression pattern into a basolateral membrane one. Using metabolic labeling and pulse-chase and surface immunoprecipitation, we found that CD4-Kir2.3 COOH-terminal chimeras were rapidly and directly targeted to the basolateral membrane, consistent with a sorting signal that is processed in the biosynthetic pathway. Collectively, the data indicate that the basolateral sorting determinant in Kir 2.3 is composed of a unique arrangement of trafficking motifs, containing tandem, conceivably overlapping, biosynthetic targeting and PDZ-based signals. The previously unrecognized domain corresponds to a highly degenerate structure within the Kir channel family, raising the possibility that the extreme COOH terminus of Kir channels may differentially coordinate membrane targeting of different channel isoforms.


Subject(s)
Potassium Channels/chemistry , Potassium Channels/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Dogs , Kidney/metabolism , Molecular Sequence Data , Mutation , Potassium Channels/metabolism , Protein Sorting Signals/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Two-Hybrid System Techniques , Tyrosine/chemistry
4.
Endocrinology ; 142(7): 2820-32, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11416001

ABSTRACT

Dopamine (DA) is a physiological regulator of PRL secretion, exerting tonic inhibitory control. DA activates an inward rectifier K(+) (IRK) channel in rat lactotropes, causing membrane hyperpolarization and inhibition of Ca(2+)-dependent action potentials. Both the activation of this effector K(+) channel and the inhibition of PRL release are mediated by D(2)-type receptor activation and pertussis toxin- sensitive G proteins. To study the molecular basis of this physiologically relevant channel, a homology-based PCR approach was employed to identify members of the IRK channel family expressed in the anterior pituitary gland. Nondegenerate primers corresponding to regions specific for IRK channels known to be G protein activated (GIRKs; gene subfamily Kir 3.0) were synthesized and used in the PCR with reverse transcribed female rat anterior pituitary messenger RNA as the template. PCR products of predicted sizes for Kir 3.1, 3.2, and 3.4 were consistently observed by ethidium bromide staining after 16 amplification cycles. The identities of the products were confirmed by subcloning and sequencing. Expression of each of these gene products in anterior pituitary was confirmed by Northern blot analysis. Functional analysis of the GIRK proteins was performed in the heterologous expression system, Xenopus laevis oocytes. Macroscopic K(+) currents were examined in oocytes injected with different combinations of Kir 3.0 complementary RNA (cRNA) and G protein subunit (beta(1)gamma(2)) cRNA. The current-voltage relationships demonstrated strong inward rectification for each individual and pairwise combination of GIRK channel subunits. Oocytes coinjected with any pair of GIRK subunit cRNA exhibited significantly larger inward K(+) currents than oocytes injected with only one GIRK channel subtype. Ligand-dependent activation of only one of the GIRK combinations (GIRK1 and GIRK4) was observed when channel subunits were coexpressed with the D(2) receptor in Xenopus oocytes. Dose-response data fit to a Michaelis-Menten equation gave an apparent K(d) similar to that for DA binding in anterior pituitary tissue. GIRK1 and GIRK4 proteins were coimmunoprecipitated from anterior pituitary lysates, confirming the presence of native GIRK1/GIRK4 oligomers in this tissue. These data indicate that GIRK1 and GIRK4 are excellent candidate subunits for the D(2)-activated, G protein-gated channel in pituitary lactotropes, where they play a critical role in excitation-secretion coupling.


Subject(s)
Pituitary Gland, Anterior/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Animals , Blotting, Northern , Electric Conductivity , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Kinetics , Oocytes , Potassium Channels/physiology , Precipitin Tests , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/physiology , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
5.
Am J Physiol Cell Physiol ; 280(1): C192-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11121391

ABSTRACT

Na(+)/H(+) exchanger regulatory factor (NHERF) and NHERF2 are PDZ motif proteins that mediate the inhibitory effect of cAMP on Na(+)/H(+) exchanger 3 (NHE3) by facilitating the formation of a multiprotein signaling complex. With the use of antibodies specific for NHERF and NHERF2, immunocytochemical analysis of rat kidney was undertaken to determine the nephron distribution of both proteins and their colocalization with other transporters and with ezrin. NHERF was most abundant in apical membrane of proximal tubule cells, where it colocalized with ezrin and NHE3. NHERF2 was detected in the glomerulus and in other renal vascular structures. In addition, NHERF2 was strongly expressed in collecting duct principal cells, where it colocalized with ROMK. These results indicate a striking difference in the nephron distribution of NHERF and NHERF2 and suggests NHERF is most likely to be the relevant biological regulator of NHE3 in the proximal tubule, while NHERF2 may interact with ROMK or other targets in the collecting duct. The finding that NHERF isoforms occur in different cell types suggests that NHERF and NHERF2 may subserve different functions in the kidney.


Subject(s)
Nephrons/metabolism , Phosphoproteins/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cytoskeletal Proteins , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/ultrastructure , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/ultrastructure , Male , Nephrons/ultrastructure , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Sodium-Hydrogen Exchanger 3
6.
J Gen Physiol ; 114(5): 685-700, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10532965

ABSTRACT

Mutations in the inward rectifying renal K(+) channel, Kir 1.1a (ROMK), have been linked with Bartter's syndrome, a familial salt-wasting nephropathy. One disease-causing mutation removes the last 60 amino acids (332-391), implicating a previously unappreciated domain, the extreme COOH terminus, as a necessary functional element. Consistent with this hypothesis, truncated channels (Kir 1.1a 331X) are nonfunctional. In the present study, the roles of this domain were systematically evaluated. When coexpressed with wild-type subunits, Kir 1.1a 331X exerted a negative effect, demonstrating that the mutant channel is synthesized and capable of oligomerization. Plasmalemma localization of Kir 1.1a 331X green fluorescent protein (GFP) fusion construct was indistinguishable from the GFP-wild-type channel, demonstrating that mutant channels are expressed on the oocyte plasma membrane in a nonconductive or locked-closed conformation. Incremental reconstruction of the COOH terminus identified amino acids 332-351 as the critical residues for restoring channel activity and uncovered the nature of the functional defect. Mutant channels that are truncated at the extreme boundary of the required domain (Kir 1.1a 351X) display marked inactivation behavior characterized by frequent occupancy in a long-lived closed state. A critical analysis of the Kir 1.1a 331X dominant negative effect suggests a molecular mechanism underlying the aberrant closed-state stabilization. Coexpression of different doses of mutant with wild-type subunits produced an intermediate dominant negative effect, whereas incorporation of a single mutant into a tetrameric concatemer conferred a complete dominant negative effect. This identifies the extreme COOH terminus as an important subunit interaction domain, controlling the efficiency of oligomerization. Collectively, these observations provide a mechanistic basis for the loss of function in one particular Bartter's-causing mutation and identify a structural element that controls open-state occupancy and determines subunit oligomerization. Based on the overlapping functions of this domain, we speculate that intersubunit interactions within the COOH terminus may regulate the energetics of channel opening.


Subject(s)
Bartter Syndrome/genetics , Frameshift Mutation , Ion Channel Gating/genetics , Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , Animals , Bartter Syndrome/physiopathology , Cell Membrane/chemistry , Cell Membrane/physiology , Electrophysiology , Female , Gene Deletion , Gene Expression/physiology , Genes, Reporter , Green Fluorescent Proteins , Humans , Indicators and Reagents , Luminescent Proteins/genetics , Membrane Potentials/genetics , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis/physiology , Oocytes/physiology , Potassium Channels/metabolism , Sequence Homology, Amino Acid , Xenopus laevis
7.
Biochem Biophys Res Commun ; 261(2): 364-71, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10425191

ABSTRACT

To investigate the biosynthetic mechanisms involved in the expression of the renal epithelial inward rectifying K(+) channel, ROMK1 (Kir1.1a), a six amino acid epitope (AU1) was introduced onto the extreme N-terminus for efficient immunoprecipitation. As expressed in Xenopus oocytes, the AU1 epitope did not modify the functional properties of the ROMK1 channel. To analyze kinetics of ROMK1 synthesis in renal epithelial cells, the AU1-ROMK1 construct was stably transfected in MDCK cells and pulse chase experiments were conducted. When the cells are grown at 37 degrees C, the ROMK1 protein was unstable, being rapidly degraded with a t(1/2) < 1 hour. Furthermore, whole cell patch clamp experiments failed to detect functional ROMK1 channels at the plasma membrane in cells grown at 37 degrees C. In contrast, the degradation process was minimized when the cells were grown at 26 degrees C (t(1/2) > 4 hours), allowing ROMK1 channels to be functionally expressed on the plasma membrane. In summary, in a mammalian epithelial expression system maintained at a physiological temperature, wild-type ROMK1 is bio-synthetically labile and incapable of efficient traffic to the plasmalemma. These observations are reminiscent of temperature sensitive biosynthetic defects in mutant plasma membrane proteins, suggesting that wild-type ROMK1 may require other factors, like the association of a surrogate subunit, for appropriate biosynthetic processing.


Subject(s)
Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Biological Transport, Active , Cell Membrane/metabolism , Dogs , Female , Gene Expression , In Vitro Techniques , Oocytes/metabolism , Potassium Channels/genetics , Protein Processing, Post-Translational , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Temperature , Transfection , Xenopus
8.
J Biol Chem ; 273(23): 14165-71, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9603917

ABSTRACT

Unique ATP-inhibitable K+ channels (KATP) in the kidney determine the rate of urinary K+ excretion and play an essential role in extracellular K+ balance. Here, we demonstrate that functionally similar low sulfonylurea affinity KATP channels are formed by two heterologous molecules, products of Kir1.1a and cystic fibrosis transmembrane conductance regulator (CFTR) genes. Co-injection of CFTR and Kir1.1a cRNA into Xenopus oocytes lead to the expression of K+ selective channels that retained the high open probability behavior of Kir1.1a but acquired sulfonylurea sensitivity and ATP-dependent gating properties. Similar to the KATP channels in the kidney but different from KATP channels in excitable tissues, the Kir1.1a/CFTR channel was inhibited by glibenclamide with micromolar affinity. Since the expression of Kir1.1a and CFTR overlap at sites in the kidney where the low sulfonylurea affinity KATP are expressed, our study offers evidence that these native KATP channels are comprised of Kir1.1a and CFTR. The implication that Kir subunits can interact with ABC proteins beyond the subfamily of sulfonylurea receptors provides an intriguing explanation for functional diversity in KATP channels.


Subject(s)
Kidney/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/chemistry , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Electrophysiology , Gene Expression/genetics , Microinjections , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/urine , Potassium Channels/genetics , RNA, Complementary/genetics , Sulfonylurea Compounds/pharmacology , Xenopus
9.
Proc Natl Acad Sci U S A ; 94(24): 13329-34, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371845

ABSTRACT

We recently cloned an inward-rectifying K channel (Kir) cDNA, CCD-IRK3 (mKir 2.3), from a cortical collecting duct (CCD) cell line. Although this recombinant channel shares many functional properties with the "small-conductance" basolateral membrane Kir channel in the CCD, its precise subcellular localization has been difficult to elucidate by conventional immunocytochemistry. To circumvent this problem, we studied the targeting of several different epitope-tagged CCD-IRK3 in a polarized renal epithelial cell line. Either the 11-amino acid span of the vesicular stomatitis virus (VSV) G glycoprotein (P5D4 epitope) or a 6-amino acid epitope of the bovine papilloma virus capsid protein (AU1) was genetically engineered on the extreme N terminus of CCD-IRK3. As determined by patch-clamp and two-microelectrode voltage-clamp analyses in Xenopus oocytes, neither tag affected channel function; no differences in cation selectivity, barium block, single channel conductance, or open probability could be distinguished between the wild-type and the tagged constructs. MDCK cells were transfected with tagged CCD-IRK3, and several stable clonal cell lines were generated by neomycin-resistance selection. Immunoprecipitation studies with anti-P5D4 or anti-AU1 antibodies readily detected the predicted-size 50-kDa protein in the transfected cells lines but not in wild-type or vector-only (PcB6) transfected MDCK cells. As visualized by indirect immunofluorescence and confocal microscopy, both the tagged CCD-IRK3 forms were exclusively detected on the basolateral membrane. To assure that the VSV G tag was not responsible for the targeting, the P5D4 epitope modified by a site-directed mutagenesis (Y2F) to remove a potential basolateral targeting signal contained in this tag. VSV(Y2F) was also detected exclusively on the basolateral membrane, confirming bona fide IRK3 basolateral expression. These observations, with our functional studies, suggest that CCD-IRK3 may encode the small-conductance CCD basolateral K channel.


Subject(s)
Kidney Tubules, Collecting/metabolism , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Basement Membrane/metabolism , Cell Line , Dogs , Epithelium/metabolism , Epitopes/chemistry , Homeostasis , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channels/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
10.
Am J Physiol ; 273(5): F825-36, 1997 11.
Article in English | MEDLINE | ID: mdl-9374848

ABSTRACT

Maintenance of a negative membrane potential in the cortical collecting duct (CCD) principal cell depends on a small-conductance, inward-rectifying basolateral membrane K+ (Kir) channel. In the present study, a candidate cDNA encoding this K+ channel, CCD-IRK3, was isolated from a mouse collecting duct cell line, M1. CCD-IRK3 shares a high degree of homology with a human brain inward-rectifier K+ channel (Kir 2.3). By Northern analysis, CCD-IRK3 transcript (2.9 kb) was readily detected in M1 CCD cells but not in Madin-Darby canine kidney, LLC-PK1, Chinese hamster ovary, or monkey kidney fibroblast cell lines. CCD-IRK3-specific reverse transcription-polymerase chain reaction confirmed bonafide expression in the kidney. Functional expression studies in Xenopus oocytes revealed that CCD-IRK3 operates as strongly inward-rectifying K+ channel. The cation selectivity profile of CCD-IRK3 [ionic permeability values (PK/Pi), Tl > or = Rb > or = K+ >> NH4 > Na; inward-slope conductance (GK/Gi), Tl > or = K+ >> NH4 > Na > Rb] is similar to the macroscopic CCD basolateral membrane K+ conductance (GK/Gi, K+ >> NH4 > Rb; PK/Pi, Rb approximately equal to K+ >> NH4). CCD-IRK3 also exhibits the pharmacological features of the native channel. Patch-clamp analysis reveals that CCD-IRK3 functions as a high open probability, voltage-independent, small-conductance channel (14.5 pS), consistent with the native channel. Based on these independent lines of evidence, CCD-IRK3 is a possible candidate for the small-conductance basolateral Kir channel in the CCD.


Subject(s)
Kidney Tubules, Collecting/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/biosynthesis , Potassium Channels/chemistry , Transcription, Genetic , Amino Acid Sequence , Animals , Anions/metabolism , Base Sequence , Brain/metabolism , CHO Cells , Cations, Monovalent/metabolism , Cell Line , Cell Membrane/physiology , Cricetinae , Dogs , Electric Conductivity , Female , Haplorhini , Humans , Ion Channel Gating , Kidney , Membrane Potentials/physiology , Mice , Molecular Sequence Data , Oocytes/physiology , Potassium Channels/physiology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus
11.
Am J Physiol ; 271(1 Pt 2): F7-15, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8760237

ABSTRACT

In the cortical collecting duct (CCD), aldosterone increases the number of functionally active Na-K-adenosin-etriphosphatase (Na-K-ATPase) molecules by a mechanism involving an isoform-specific increase in the abundance of the Na-K-ATPase alpha 1- and beta 1-subunit protein. However, the molecular basis for the response, particularly in the mammalian CCD in vivo, has remained unclear. To resolve this issue, reverse transcription (RT) and a competitive polymerase chain reaction (PCR) were employed to study mineralocorticoid-dependent regulation of alpha 1- and beta 1-subunit mRNA in the rat CCD. Na-K-ATPase subunit-specific oligonucleotides primers were used in the PCR to amplify reverse-transcribed subunit mRNA (RT-mRNA) from single microdissected CCD. Control templates were constructed (84-bp deletion mutation of the rat Na-K-ATPase alpha 1-subunit cDNA and 70-bp deletion of the beta 1-subunit cDNA), serially diluted, and coamplified with the wild-type Na-K-ATPase subunit RT-mRNA from single CCD. PCR products of predicted size were observed by ethidium bromide staining. Southern blots with an internal subunit-specific oligonucleotide confirmed Na-K-ATPase alpha 1- and beta 1-subunit identity. The ratio of the amplified wild-type to mutant PCR products was found to be linear over the range of input control cDNA tested so that the amount of subunit mRNA could be determined. A chronic reduction in corticosteroid levels by bilateral adrenalectomy (7 days) reduced the apparent level of alpha 1-subunit transcript by 54.0 +/- 6.3% but not the beta 1-subunit. Administering aldosterone to physiological levels is sufficient to restore CCD alpha 1-subunit mRNA abundance toward control levels within 6 h. We conclude the following: 1) regulation of Na-K-ATPase of CCD in vivo can be attributed, at least in part, to mineralocorticoid-dependent control of Na-K-ATPase alpha 1-subunit mRNA abundance; and 2) competitive PCR may provide a sensitive and quantitative tool for determining hormone-dependent regulation of mRNA abundance in nephron segments.


Subject(s)
Aldosterone/physiology , Isoenzymes/genetics , Kidney Tubules, Collecting/metabolism , RNA, Messenger/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Adrenalectomy , Aldosterone/pharmacology , Animals , Female , Kidney Cortex , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
12.
Am J Physiol ; 270(4 Pt 1): C1246-54, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8928752

ABSTRACT

A phenomenological model is presented of water and solute transport that is applicable to water pores with radii less than approximately 2 A. This includes such examples as gramicidin A, the proximal tubule basolateral membrane, and the aquaporin 1 (CHIP28) water channel. The model differs from the conventional single-file model by allowing for a variation of unoccupied volume within the pores. It is shown that the accessible or free portion of the unoccupied volume can be related to the mechanical frictional coefficients and thereby to the filtration and diffusive permeabilities by the filled pore approximation. In general, the smallness of the unoccupied volume represents the compactness of the molecules within the pore and is indicative of the single-file character of the motion of water and solute moving together. When that volume is equal to a single water volume, the results are identical to the conventional single-file model. An important result is that, despite very low diffusive permeabilities, the reflection coefficient of a solute can remain at approximately 0.5 if its frictional interaction with the channel walls is comparable with its frictional interaction with neighboring water molecules. This is consistent with values previously reported for NaCl in cell membranes of proximal tubule. The model predicts a minimum effective pore radius for a water channel of 1.78 A and corresponds to a maximum filtration-to-diffusion permeability ratio that is proportional to the length of the effective pore or channel. This limiting condition corresponds to a water channel completely filled by water and may be applicable to the aquaporin 1 water channel.


Subject(s)
Aquaporins , Ion Channels/physiology , Models, Biological , Animals , Aquaporin 1 , Blood Group Antigens , Cell Membrane/metabolism , Gramicidin/pharmacology , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism
13.
Kidney Int ; 48(4): 1017-23, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8569062

ABSTRACT

Over the last few years it has become evident that an assortment of functionally-related, but diverse, KATP channels provide an important and physiologically-regulated determinant of the K conductive pathways in many, if not all, epithelial cells expressed along the nephron. As such, KATP plays central roles in regulating and maintaining a number of transport processes in concert with physiological demands of the kidney. In the renal proximal tubule, KATP channels and changes in the hydrolytic activity of the Na,K-ATPase permit ATP to act as a coupling modulator of parallel Na,K-ATPase-K recycling. The response insures that cell membrane potential, intracellular K activity and cell volume are protected in the face of physiological variations in transcellular ion transport. In addition to demonstrating the physiological relevance of KATP in renal epithelial, these studies have provided a long awaited answer to the underlying mechanism of pump-leak coupling, a universal and essential homeostatic mechanism observed in nearly all salt translocating epithelia.


Subject(s)
Adenosine Triphosphate/metabolism , Kidney Tubules, Proximal/metabolism , Potassium Channels/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cell Size , Humans , Hydrogen-Ion Concentration , Models, Biological , Molecular Biology , Sodium-Potassium-Exchanging ATPase/metabolism
14.
J Biol Chem ; 268(31): 23469-76, 1993 Nov 05.
Article in English | MEDLINE | ID: mdl-8226873

ABSTRACT

In the renal cortical collecting duct (CCD), mineralocorticoid hormones, like aldosterone, augment the abundance of Na/K-ATPase molecules. It has been postulated that this response involves an isoform switch of the Na/K-ATPase catalytic subunit, alpha, as the molecular basis for the differential regulation of mineralo-corticoid-induced and constitutively expressed Na/K-ATPase pools. In opposition to this attractive hypothesis, three lines of independent evidence are presented which demonstrate that the CCD exclusively expresses the alpha 1 form despite mineralocorticoid-mediated changes in functional Na/K pump density. First, aldosterone increased [3H]ouabain binding in CCD 2.5-fold without changing the ouabain dissociation constant. Second, an electrophysiological assay for pump activity revealed that aldosterone increased maximum Na/K pump current in parallel with the change in ouabain binding without altering the apparent sodium affinity. Third, Western blot analysis with alpha isoform-specific, antipeptide antibodies demonstrated that aldosterone exclusively increased the total chemical pool of the alpha 1 form of the pump without inducing other alpha subunit isoforms. In summary, aldosterone increases the abundance of Na/K-ATPase molecules in the CCD which are pharmacologically, physiologically, and chemically indistinguishable from those that are normally expressed.


Subject(s)
Aldosterone/pharmacology , Kidney Tubules, Collecting/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blotting, Western , Female , Isoenzymes/metabolism , Kinetics , Ouabain/metabolism , Rabbits
15.
Proc Natl Acad Sci U S A ; 89(14): 6418-22, 1992 Jul 15.
Article in English | MEDLINE | ID: mdl-1321439

ABSTRACT

A fundamental and essential property of nearly all salt-transporting epithelia is the tight parallel coupling between the magnitude of the K-conductive pathway at the basolateral membrane and the activity of the Na,K-dependent ATPase (Na,K-ATPase). In the present study, we demonstrate that the coupling response in the renal proximal tubule is governed, at least in part, through the interaction between ATP-sensitive K channels and Na,K-ATPase-mediated changes in intracellular ATP levels. First, we identified a K-selective channel at the basolateral membrane, which is inhibited by the cytosolic addition of ATP. Second, conventional microelectrode analysis in the isolated perfused proximal straight tubule revealed that these channels are the major determinant of the macroscopic K conductance so that ATP-mediated changes in the open probability of the K channel could alter the extent of K recycling. Indeed, the increase in the macroscopic K conductance upon stimulation of transcellular Na transport and pump activity was found to be paralleled by a decrease in intracellular ATP. Finally, a causal link between parallel Na,K-ATPase-K-channel activity and ATP was established by the finding that intracellular ATP loading uncoupled the response. With our recent observations that similar ATP-sensitive K channels are expressed abundantly in other epithelia, we postulate that ATP may act as a universal coupling modulator of parallel Na,K-ATPase-K-channel activity.


Subject(s)
Adenosine Triphosphate/physiology , Kidney Tubules, Proximal/physiology , Potassium Channels/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Glyburide/pharmacology , In Vitro Techniques , Ion Channel Gating , Membrane Potentials/drug effects , Potassium/metabolism , Rabbits , Signal Transduction , Sodium/metabolism
16.
Am J Physiol ; 258(4 Pt 2): F951-62, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2330988

ABSTRACT

The ionic basis of volume regulation was assessed in the nonperfused rabbit proximal tubule (S2 segment) by use of simultaneous measurements of tubule volume via video-optical imaging techniques and basolateral membrane voltage (Vbl) and relative ionic conductance via conventional microelectrodes. Both cell volume (9.9 +/- 0.70 nl/cm tubule length) and Vbl (-42.8 +/- 3.6 mV) remained stable in the control isotonic Ringer solution (290 mosmol/kg). When the osmolality of the bathing medium was reduced to 150 mosmol/kg, tubules swelled 72% above base line within 1 min and subsequently regulated over the course of the next 4-6 min to a steady state 20 +/- 3% above the initial base-line volume. Cell swelling was accompanied by a transient hyperpolarization of Vbl of -14.3 +/- 2.0 mV (HCO3-containing Ringer) and -10.0 +/- 0.7 mV (HCO3-free Ringer). Although the hyperpolarization was not inhibited by barium in the presence of bicarbonate buffer, addition of 2 mM Ba to a bicarbonate-free Ringer depolarized Vbl by +22 mV and abolished both the relative potassium conductance and the hyperpolarization accompanying cell swelling (delta Vbl = -4.6 +/- 0.6 mV). Furthermore, the relative conductance of K at the basolateral membrane increased from 0.16 in the isotonic control medium to 0.34 at the peak of cell swelling. Because the hyperpolarization of Vbl ensued after cells had swollen approximately 10% above base line, a modest threshold volume and time delay may be involved in triggering the volume-dependent activation of the K conductance. In parallel studies, the change in Vbl on a rapid step-change in bath Cl (49 to 4.9 mM) averaged 5.3 +/- 1.0 mV in the isotonic solution and increased to +11.3 +/- 2.1 (P less than or equal to 0.05) at the peak of cell swelling. This represented an increase in the relative Cl conductance of 0.08 to 0.20, which could only be attributed to an absolute increase in the basolateral membrane Cl conductance and not to a reduction in the other major basolateral membrane conductances. It is concluded that cell swelling results in an increase in both Cl and K conductance, which may underlie subsequent cell volume regulation.


Subject(s)
Chlorides/pharmacokinetics , Intracellular Membranes/metabolism , Kidney Tubules, Proximal/metabolism , Potassium/pharmacokinetics , Animals , Barium/pharmacology , Bicarbonates/metabolism , Biological Transport , Carrier Proteins/metabolism , Electrophysiology/instrumentation , Hypotonic Solutions/pharmacology , In Vitro Techniques , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/physiology , Osmolar Concentration , Rabbits , Sodium Chloride/pharmacology , Sodium-Bicarbonate Symporters , Time Factors
17.
Am J Physiol ; 258(4 Pt 2): F940-50, 1990 Apr.
Article in English | MEDLINE | ID: mdl-2330987

ABSTRACT

The ionic conductive properties of the nonperfused rabbit proximal straight tubule (S2) basolateral membrane were assessed by microelectrode techniques. The response of the basolateral membrane electrical potential difference, Vbl, to rapid changes in the peritubular bath concentration of K, HCO3, Na, and Cl were monitored with microelectrodes. The control steady-state Vbl averaged -41 mV (cell negative). An increase in peritubular bathing medium K concentration from 5 to 40 mM resulted in an instantaneous and sustained depolarization of +14.6 mV (27% of delta EK). Addition of barium (2 mM) depolarized the Vbl by +15.8 mV and abolished the Vbl response to the high-K medium. In other studies, reduction of peritubular bicarbonate at constant pH from 25 to 2.5 mM instantaneously and transiently depolarized Vbl by +15.8 mV (26% of delta EHCO3). In these same tubules reduction of peritubular Na from 126 to 2.2 mM resulted in an instantaneous and paradoxical depolarization of Vbl of +21.5 mV. Both depolarization transients resulting from reduction of Na and HCO3 were simultaneously inhibited by the addition of 4-acetamido-4'-isothiocyanostilbene-2,2'-disulfonic acid (SITS; 0.5 mM), consistent with the presence of a SITS-sensitive Na-HCO3-coupled conductive pathway. In the absence of the bicarbonate buffer, reduction of Na resulted in a small sustained hyperpolarization of -5.8 mV (5% of delta ENa). Reduction of peritubular Cl from 120 to 4 mM resulted in an instantaneous and sustained depolarization of Vbl of +5.3 mV (6% of ECl) and was not affected by the addition of bumetanide (0.1 mM). It is concluded that the basolateral membrane of the nonperfused proximal straight tubule is characterized by a major barium-sensitive K conductance and a SITS-sensitive Na-coupled HCO3 conductance that carries net negative charge. These pathways are paralleled by relatively minor, but important, Na-conductive and Cl-conductive pathways.


Subject(s)
Kidney Tubules, Proximal/metabolism , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Animals , Bicarbonates/pharmacology , Chlorides/pharmacokinetics , Electrophysiology , Female , Ions , Kidney Tubules, Proximal/physiology , Membrane Potentials , Membranes/metabolism , Membranes/physiology , Potassium/pharmacokinetics , Rabbits , Sodium/pharmacokinetics , Sodium/pharmacology
18.
Am J Physiol ; 255(5 Pt 2): F853-60, 1988 Nov.
Article in English | MEDLINE | ID: mdl-3189560

ABSTRACT

Rabbit proximal straight tubules rapidly swell to a maximum volume when abruptly immersed into hypotonic medium. However, in a second, slower phase, termed volume regulatory decrease (VRD), tubules shrink toward their basal volume due to the efflux of K, accompanying anion and water. In the present study, we investigated the nature of the anion during hypotonic volume regulation. We removed Cl and/or the HCO3 buffer to assess their relative importance in VRD, and we used furosemide, 4-acetamido-4'-isothiocyanostilbene-2,2'-disulfonic acid (SITS), and barium to investigate the nature of the transmembrane anion pathway in VRD. Isosmotic replacement of peritubular Cl with gluconate had no effect on either tubule volume in isotonic medium or the initial osmometric swelling response in hypotonic medium. However, the VRD in such Cl-depleted tubules was significantly inhibited. Control tubules regulated 81% below their maximal volume in dilute medium. By contrast, Cl-depleted tubules regulated only 39%. This inhibitory effect could not be attributed to the absence of peritubular Cl or to the presence of gluconate. The absence of the HCO3 buffer or the presence of SITS (0.5 mM) had no inhibitory effect on the rate or extent of VRD. Furosemide alone (1 mM) also had no inhibitory effect on VRD. However, whereas barium alone delays VRD, addition of furosemide to barium-treated tubules further slowed their maximal rate of fluid efflux and delayed VRD even more.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Chlorides/metabolism , Kidney Tubules, Proximal/physiology , Water-Electrolyte Balance , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid , Animals , Anions , Barium/pharmacology , Bicarbonates/metabolism , Cell Membrane/metabolism , Female , Furosemide/pharmacology , Hypotonic Solutions , Kidney Tubules, Proximal/anatomy & histology , Kidney Tubules, Proximal/drug effects , Kinetics , Potassium/metabolism , Rabbits , Water-Electrolyte Balance/drug effects
19.
Am J Physiol ; 249(1 Pt 2): F20-7, 1985 Jul.
Article in English | MEDLINE | ID: mdl-4014473

ABSTRACT

Rabbit proximal straight tubules swell abruptly when exposed to hypotonic medium but then shrink in a few minutes as they approach their base-line volume following loss of solute and water. Potassium, the major intracellular cation, as well as sodium, is lost during this process. In the present experiments, we studied hypotonic cell volume regulation in the presence of barium, an agent reported to decrease potassium permeability. Exposure to BaCl2 significantly prolonged hypotonic volume recovery in a dose-dependent manner. Tubules depleted of potassium and loaded with sodium chloride by exposure to 10(-4) M ouabain for 1 h swelled osmometrically and subsequently volume regulated in dilute medium. Volume regulation in such tubules is a consequence of transbasement membrane hydrostatic forces. By contrast, tubules similarly loaded with sodium, but also exposed to 10(-3) M BaCl2, volume regulated only minimally in dilute medium, suggesting BaCl2 might also affect sodium movement. However, hypotonic volume regulation was restored in sodium-loaded BaCl2-treated tubules when cells were more effectively depleted of potassium by incubation in 0-mM potassium medium. We conclude that barium retards hypotonic volume regulation primarily because of its effect on potassium movement.


Subject(s)
Barium Compounds , Barium/pharmacology , Chlorides , Kidney Tubules, Proximal/metabolism , Potassium/metabolism , Water-Electrolyte Balance/drug effects , Animals , Basement Membrane/metabolism , Biological Transport, Active/drug effects , Cell Membrane Permeability/drug effects , Culture Media , Dose-Response Relationship, Drug , Female , Hydrostatic Pressure , Kidney Tubules, Proximal/drug effects , Osmolar Concentration , Ouabain/pharmacology , Rabbits , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...